• The EHA-IDWP developed an observational registry collecting data on COVID-19 infection in patients who received CAR T-cell therapy.

  • Prevalence of COVID-19 was 4.8%, and overall mortality was 50%, highlighting the need for prevention of infection in these patients.

Since it was first reported in China, coronavirus disease 2019 (COVID-19) has spread rapidly around the world, and the number of cases has increased exponentially.1  Initial reports suggested that patients with cancer have an estimated twofold increased risk of contracting severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) compared with the general population.2  More importantly, it is expected that COVID-19 will be particularly life threatening in patients with hematological malignancies because of their immune dysfunction. Recent studies have reported an overall COVID-19–related mortality of 29% to 42%3-8  in patients with hematological disease, depending on the type of malignancy, in contrast to the 2% to 7% observed in the general population. Regrettably, there remains a lack of studies about COVID-19 in patients receiving cellular therapies, including chimeric antigen receptor (CAR) T cells.9,10  CAR T cells are genetically modified autologous T cells, which have shown great promise in the treatment of advanced malignant hematological disorders, including non-Hodgkin lymphoma, acute lymphoblastic leukemia, and multiple myeloma.11  CAR T-cell recipients have significant B-cell aplasia requiring immunoglobulin G replacement therapy and may also develop delayed cytopenias, leaving them unable to mount any humoral response to viral infections.12  Shah et al10  demonstrated that the seroconversion rate in a small cohort of patients treated with hemopoietic stem cell transplantation (HSCT) and CAR T-cell therapy did not exceed 66%.

According to these observations, the outcomes of COVID-19 in patients treated with CAR T cells remain unclear. The aim of this study was to describe the clinical outcomes of patients developing COVID-19 after treatment with CAR T cells.

In this retrospective observational multicenter study, we collected data on all consecutive adult patients who received CAR T-cell therapy with symptomatic COVID-19 between January 2020 and February 2021 across 18 European centers (Spain, n = 6; France, n = 3; Italy, n = 2; and Belgium, Croatia, Czechia, Slovakia, Sweden, Switzerland, and the United Kingdom, n = 1 each) participating in the survey promoted by the European Hematology Association (EHA) Scientific Working Group on Infection in Hematology (EPICOVIDEHA survey),13  developed by the EHA Infectious Diseases Working Party (IDWP). Confirmed cases of COVID-19 were defined by positive reverse transcription polymerase chain reaction assays of specimens collected on nasopharyngeal swabs. Each institutional review board independently approved the study. The study was conduct in accordance with the Declaration of Helsinki. Researchers at each center collected data using an online questionnaire hosted at www.clinicalsurveys.net. The EPICOVIDEHA trial was registered at www.clinicaltrials.gov as #NCT04733729. Only deidentified data have been entered and analyzed. We obtained demographic data, comorbidities, and underlying hematological disease, including clinically significant outcomes (hospital and intensive care unit [ICU] admission and vital status) and management strategies of COVID-19. The severity of COVID-19 at admission was graded according to the China Centers for Disease Control and Prevention definitions: mild (nonpneumonia and mild pneumonia), severe (dyspnea, respiratory frequency ≥30 breaths per minute, oxygen saturation ≤93%, or ratio of arterial oxygen partial pressure/fractional inspired oxygen of 50%), and critical (respiratory failure, septic shock, or multiple organ dysfunction or failure).

SPSS v25.0 was employed for statistical analyses (IBM Corp., Chicago, IL). Categorical variables are presented using frequency and percentage, and continuous variables are shown by median, interquartile range (IQR), and absolute range. Additionally, overall mortality was evaluated by employing a Cox proportional hazards model. Univariable Cox regression model was performed with variables suspected to play a role in the mortality of patients with COVID-19 receiving CAR T-cell therapy. Variables with a P value ≤.1 were considered for multivariable analysis. Multivariable Cox regression model was calculated with the Wald backward method, and only statistically significant variables were reported. A P value ≤.05 was considered statistically significant.

Overall, 459 patients received CAR T-cell therapy, of whom 30 met the criteria for diagnosis of COVID-19. Median age at COVID-19 diagnosis was 57 years (IQR, 51-64; range, 18-74); 13 patients (43.3%) were female, and 17 patients (56.7%) were male. Demographic and clinical characteristics of CAR T-cell recipients with COVID-19 are summarized in Table 1. Patients received CAR T cells for the treatment of large B-cell lymphoma (n = 28), multiple myeloma (n = 1), and acute lymphoblastic leukemia (n = 1). A majority of patients received CAR-T therapy in 2020 (n = 17), 3 in 2018, 9 in 2019, and 1 in 2021. CAR T cells were tisagenlecleucel (Kymriah) in 16 cases and axicabtagene (Yescarta) in 13 cases, and 1 patient with multiple myeloma was treated with CAR T cells targeting B-cell maturation antigen. A majority of patients received bridging therapy (n = 21 of 30) and fludarabine plus cyclophosphamide as lymphodepletion conditioning (n = 29 of 30). Severe (grade ≥3) CRS after receiving CAR T cells was observed in 1 patient only. No patient received COVID-19 vaccine. Seventeen patients (56.7%) developed COVID-19 within 6 months from CAR T-cell infusion, and 13 patients (43.3%) developed it after 6 months. Prevalence of COVID-19 in our patients was 4.8%, based on the total number of CAR T-cell recipients reported by participating centers in 2020 (n = 17 of 353).

Table 1.

Characteristics of patients with COVID-19 receiving CAR T-cell therapy (n = 30)

PatientBaseline
hematological malignancy
Disease status at COVID-19 diagnosisTime from CAR T cells to COVID-19, dCAR T-cell constructBridging therapyLymphodepletionGrade at CAR T-cell therapyCRS/ICANS treatmentReason for death
(time after
COVID-19 diagnosis, d)*
CRSICANSTocilizumabSteroids
NHL LBCL R/R 37 Axi-cel Flu-Cyc   Malignancy (35) 
NHL LBCL CR 221 Axi-cel Flu-Cyc NA   NA 
NHL LBCL CR 240 Axi-cel  Flu-Cyc NA COVID-19 + other (52) 
NHL LBCL R/R Tisa-cel Flu-Cyc NA COVID-19 (25) 
NHL LBCL CR 85 Axi-cel  Flu-Cyc  NA 
NHL LBCL SD 14 Tisa-cel Benda COVID-19 + other (170) 
MM R/R 90 BCMA ARI-0002 Flu-Cyc NA NA   NA 
NHL LBCL R/R 42 Tisa-cel Flu-Cyc NA Malignancy + COVID-19 (33) 
NHL LBCL R/R 397 Tisa-cel Flu-Cyc NA  NA 
10 NHL LBCL CR 313 Axi-cel Flu-Cyc NA  NA 
11 ALL Unknown Tisa-cel  Flu-Cyc NA NA   NA 
12 NHL LBCL R/R 302 Axi-cel Flu-Cyc NA  COVID-19 (42) 
13 NHL LBCL CR 95 Tisa-cel Flu-Cyc NA NA   NA 
14 NHL LBCL CR 457 Axi-cel Flu-Cyc NA   NA 
15 NHL LBCL R/R 158 Axi-cel Flu-Cyc NA Malignancy (22) 
16 NHL LBCL R/R 65 Tisa-cel Flu-Cyc Malignancy + COVID-19 (21) 
17 NHL LBCL CR 270 Axi-cel Flu-Cyc NA NA 
18 NHL LBCL Unknown 19 Tisa-cel  Flu-Cyc NA  COVID-19 (50) 
19 NHL LBCL R/R 180 Tisa-cel  Flu-Cyc NA NA 
20 NHL LBCL CR 186 Tisa-cel  Flu-Cyc NA   COVID-19 (51) 
21 NHL LBCL CR 348 Axi-cel  Flu-Cyc NA 
22 NHL LBCL CR 19 Tisa-cel Flu-Cyc NA   NA 
23 NHL LBCL CR 605 Tisa-cel Flu-Cyc NA NA   COVID-19 (46) 
24 NHL NOS CR 635 Axi-cel  Flu-Cyc NA   COVID-19 (44) 
25 NHL LBCL R/R 180 Tisa-cel Flu-Cyc NA   Malignancy + COVID-19 (32) 
26 NHL LBCL CR 485 Axi-cel Flu-Cyc NA   COVID-19 (53) 
27 NHL LBCL CR 510 Axi-cel  Flu-Cyc NA   COVID-19 (124) 
28 NHL LBCL CR 66 Tisa-cel Flu-Cyc NA  NA 
29 NHL LBCL R/R 12 Tisa-cel Flu-Cyc   NA 
30 NHL LBCL CR 35 Tisa-cel Flu-Cyc NA NA   NA 
PatientBaseline
hematological malignancy
Disease status at COVID-19 diagnosisTime from CAR T cells to COVID-19, dCAR T-cell constructBridging therapyLymphodepletionGrade at CAR T-cell therapyCRS/ICANS treatmentReason for death
(time after
COVID-19 diagnosis, d)*
CRSICANSTocilizumabSteroids
NHL LBCL R/R 37 Axi-cel Flu-Cyc   Malignancy (35) 
NHL LBCL CR 221 Axi-cel Flu-Cyc NA   NA 
NHL LBCL CR 240 Axi-cel  Flu-Cyc NA COVID-19 + other (52) 
NHL LBCL R/R Tisa-cel Flu-Cyc NA COVID-19 (25) 
NHL LBCL CR 85 Axi-cel  Flu-Cyc  NA 
NHL LBCL SD 14 Tisa-cel Benda COVID-19 + other (170) 
MM R/R 90 BCMA ARI-0002 Flu-Cyc NA NA   NA 
NHL LBCL R/R 42 Tisa-cel Flu-Cyc NA Malignancy + COVID-19 (33) 
NHL LBCL R/R 397 Tisa-cel Flu-Cyc NA  NA 
10 NHL LBCL CR 313 Axi-cel Flu-Cyc NA  NA 
11 ALL Unknown Tisa-cel  Flu-Cyc NA NA   NA 
12 NHL LBCL R/R 302 Axi-cel Flu-Cyc NA  COVID-19 (42) 
13 NHL LBCL CR 95 Tisa-cel Flu-Cyc NA NA   NA 
14 NHL LBCL CR 457 Axi-cel Flu-Cyc NA   NA 
15 NHL LBCL R/R 158 Axi-cel Flu-Cyc NA Malignancy (22) 
16 NHL LBCL R/R 65 Tisa-cel Flu-Cyc Malignancy + COVID-19 (21) 
17 NHL LBCL CR 270 Axi-cel Flu-Cyc NA NA 
18 NHL LBCL Unknown 19 Tisa-cel  Flu-Cyc NA  COVID-19 (50) 
19 NHL LBCL R/R 180 Tisa-cel  Flu-Cyc NA NA 
20 NHL LBCL CR 186 Tisa-cel  Flu-Cyc NA   COVID-19 (51) 
21 NHL LBCL CR 348 Axi-cel  Flu-Cyc NA 
22 NHL LBCL CR 19 Tisa-cel Flu-Cyc NA   NA 
23 NHL LBCL CR 605 Tisa-cel Flu-Cyc NA NA   COVID-19 (46) 
24 NHL NOS CR 635 Axi-cel  Flu-Cyc NA   COVID-19 (44) 
25 NHL LBCL R/R 180 Tisa-cel Flu-Cyc NA   Malignancy + COVID-19 (32) 
26 NHL LBCL CR 485 Axi-cel Flu-Cyc NA   COVID-19 (53) 
27 NHL LBCL CR 510 Axi-cel  Flu-Cyc NA   COVID-19 (124) 
28 NHL LBCL CR 66 Tisa-cel Flu-Cyc NA  NA 
29 NHL LBCL R/R 12 Tisa-cel Flu-Cyc   NA 
30 NHL LBCL CR 35 Tisa-cel Flu-Cyc NA NA   NA 

Other reasons for death: Patient #3, possible IFI; patient #6, pulmonary embolism and heart failure.

ALL, acute lymphoblastic leukemia; axi-cel, axicabtagene ciloleucel; BCMA, B-cell maturation antigen; benda, bendamustine; CR, complete remission; CRS, cytokine release syndrome; Flu-Cyc, fludarabine plus cyclophosphamide; ICANS, immune effector cell-associated neurotoxicity syndrome; LBCL, large B-cell lymphoma; MM, multiple myeloma; NA, not applicable; NHL, non-Hodgkin lymphoma; R/R, relapsed/refractory; SD, stable disease; tisa-cel, tisagenlecleucel.

*

Other reasons for death: patient 3, possible invasive fungal infection; patient 6, pulmonary embolism and heart failure.

Median time from CAR T-cell treatment to COVID-19 diagnosis was 169 days (IQR, 37-313; range, 1-635). Cellular and humoral immune reconstitution after CAR T cells showed that 90 days after infusion, median absolute neutrophil count and absolute lymphocyte count (ALC) were 1700/mm3 (IQR, 1090-2700; range, 300-11 260) and 435/mm3 (IQR, 200-775; range, 80-3500), respectively, whereas at the time of COVID-19 diagnosis, median absolute neutrophil count and ALC were 925/mm3 (IQR, 495-2450; range, 18-11 510) and 370/mm3 (IQR, 200-1250; range, 6-1750), respectively. Overall, 10% (n = 3), 20% (n = 6), and 66.7% (n = 20) of patients had asymptomatic, mild, or severe COVID-19, respectively. Comorbidities preceding diagnosis of COVID-19 were detected in 19 patients (76.7%), including chronic cardiopathy (n = 8; 23.3%), chronic pulmonary diseases (n = 7; 23.3%), smoking history (n = 6; 20.0%), and obesity (n = 5; 16.7%). In total, 13 patients (43.3%) required admission to ICU after COVID-19, and 9 of them (66.7%) required mechanical ventilation.

Patients received treatment for COVID-19 according to local policy; 15 patients were treated with convalescent plasma alone (n = 3) or convalescent plasma combined with remdesivir with or without steroids (n = 8), remdesivir with lopinavir/ritonavir and steroids (n = 1), and tocilizumab and steroids (n = 3); 5 patients were treated with steroids alone (n = 4) or steroids combined with remdesivir and tocilizumab (n = 1); 1 patient was treated with azithromycin; and 1 patient was treated with remdesivir alone. In total, 5 patients did not require any specific treatment; in 2 cases, this was because of poor general condition, and in 3 cases, treatment was unknown.

Median follow-up was 71 days (IQR, 44-142; range, 21-379) after CAR T-cell infusion, and at the last follow-up, 15 patients (50.0%) were alive and 15 patients (50.0%) had died. Ten patients (33%) died as a result of COVID-19 infection (associated with pulmonary embolism plus heart failure in 1 case and possible fungal infection in 1 case), and 5 patients died as a result of recurrent underlying disease (associated with COVID-19 infection in 3 patients). Seven (63.6%) of the 11 patients with relapsed/refractory disease and 7 (41.1%) of the 17 patients with complete remission/stable disease died as a result of COVID-19. In 2 patients, baseline malignancy status was unknown; 1 patient died, and 1 survived.

Severe (grade ≥3) CRS after CAR T cells was observed in 1 patient only, and in total, 8 (53.3%) of 15 patients who developed CRS after CAR T-cell infusion required treatment with tocilizumab with or without steroids. None of the parameters analyzed in the univariable analysis had a significant impact on patient outcomes. Only disease status at the time of COVID-19 was marginally significant for adverse outcome (P = .075), adjusted by sex (male vs female; P = .0092; Table 2).

Table 2.

Univariable and multivariable analyses of factors associated with mortality of patients with COVID-19 receiving CAR T cells

All patientsTime between CAR T-cell therapy and COVID-19, mo
≤6>6
UnivariableMultivariableUnivariableUnivariable
HR95% CIPHR95% CIPHR95% CIPHR95% CIP
Sex             
 Female — — — — — — — — — — — — 
 Male 2.682 0.849-8.474 .093 2.742 0.848-8.861 .092 4.897 0.591-40.604 .141 1.928 0.417-8.908 .401 
Age, y             
 <50 — — — — — — — — — — — — 
 ≥50 5.119 0.673-38.955 .115 — — — 1.809 0.222-14.742 .580 42.159 0.068-26 050.639 .254 
Comorbidities, n             
 None — — — — — — — — — — — — 
 1 3.093 0.772-12.393 .111 — — — 3.093 0.772-12.393 .111 3.438 0.482-24.537 .218 
 2 3.021 0.498-18.328 .229 — — — 3.021 0.498-18.328 .229 2.055 0.185-22.871 .558 
 ≥3 1.880 0.413-8.562 .414 — — — 1.880 0.413-8.562 .414 6.111 0.820-45.529 .077 
Malignancy status at COVID-19 diagnosis             
 Controlled disease — — — — — — — — — — — — 
 Active disease 2.707 0.931-7.870 .067 2.652 0.907-7.754 .075 2.707 0.931-7.870 .944 1.121 0.133-9.446 .916 
 Unknown 1.579 0.188-13.238 .674 1.059 0.123-9.132 .958 — — .947 — — — 
CAR T-cell construct             
 Axi-cel — — — — — — — — — — — — 
 Tisa-cel 0.888 0.321-2.458 .820 — — — 0.479 0.092-2.494 .382 1.552 0.296-8.132 .603 
 Other — — .986 — — — — — .991 — — — 
ICU stay 1.529 0.554-4.225 .413 — — — 0.887 0.211-3.729 .870 3.331 0.643-17.247 .152 
Tocilizumab/steroids after CAR T cells 1.437 0.520-3.972 .484 — — —       
Time from CAR T cells to COVID-19, mo             
 ≤6 — — — — — —       
 >6 0.998 0.359-2.770 .996 — — —       
Neutrophils at COVID-19 diagnosis, n per mm3             
 ≤500 — — — — — — — — — — — — 
 >500 0.611 0.161-2.321 .469 — — — 0.761 0.167-3.472 .724 — — — 
Lymphocytes at COVID-19 diagnosis, n per mm3             
 ≤200 — — — — — — — — — — — — 
 >200 0.551 0.164-1.846 .334 — — — 0.568 0.133-2.419 .444 0.872 0.089-8.511 .907 
All patientsTime between CAR T-cell therapy and COVID-19, mo
≤6>6
UnivariableMultivariableUnivariableUnivariable
HR95% CIPHR95% CIPHR95% CIPHR95% CIP
Sex             
 Female — — — — — — — — — — — — 
 Male 2.682 0.849-8.474 .093 2.742 0.848-8.861 .092 4.897 0.591-40.604 .141 1.928 0.417-8.908 .401 
Age, y             
 <50 — — — — — — — — — — — — 
 ≥50 5.119 0.673-38.955 .115 — — — 1.809 0.222-14.742 .580 42.159 0.068-26 050.639 .254 
Comorbidities, n             
 None — — — — — — — — — — — — 
 1 3.093 0.772-12.393 .111 — — — 3.093 0.772-12.393 .111 3.438 0.482-24.537 .218 
 2 3.021 0.498-18.328 .229 — — — 3.021 0.498-18.328 .229 2.055 0.185-22.871 .558 
 ≥3 1.880 0.413-8.562 .414 — — — 1.880 0.413-8.562 .414 6.111 0.820-45.529 .077 
Malignancy status at COVID-19 diagnosis             
 Controlled disease — — — — — — — — — — — — 
 Active disease 2.707 0.931-7.870 .067 2.652 0.907-7.754 .075 2.707 0.931-7.870 .944 1.121 0.133-9.446 .916 
 Unknown 1.579 0.188-13.238 .674 1.059 0.123-9.132 .958 — — .947 — — — 
CAR T-cell construct             
 Axi-cel — — — — — — — — — — — — 
 Tisa-cel 0.888 0.321-2.458 .820 — — — 0.479 0.092-2.494 .382 1.552 0.296-8.132 .603 
 Other — — .986 — — — — — .991 — — — 
ICU stay 1.529 0.554-4.225 .413 — — — 0.887 0.211-3.729 .870 3.331 0.643-17.247 .152 
Tocilizumab/steroids after CAR T cells 1.437 0.520-3.972 .484 — — —       
Time from CAR T cells to COVID-19, mo             
 ≤6 — — — — — —       
 >6 0.998 0.359-2.770 .996 — — —       
Neutrophils at COVID-19 diagnosis, n per mm3             
 ≤500 — — — — — — — — — — — — 
 >500 0.611 0.161-2.321 .469 — — — 0.761 0.167-3.472 .724 — — — 
Lymphocytes at COVID-19 diagnosis, n per mm3             
 ≤200 — — — — — — — — — — — — 
 >200 0.551 0.164-1.846 .334 — — — 0.568 0.133-2.419 .444 0.872 0.089-8.511 .907 

Axi-cel, axicabtagene ciloleucel; CI, confidence interval; HR, hazard ratio; tisa-cel, tisagenlecleucel.

In the present multicenter international study, we sought to evaluate COVID-19 outcomes in a cohort of 459 consecutive patients who received CAR T-cell therapy. Several studies have addressed clinical courses and outcomes of COVID-19 in patients with hematological disease as well as the presence of risk factors for more aggressive life-threatening disease.4,7,14-16  Patients with hematological malignancies may be considered more vulnerable than the general population; however, the exact prevalence of SARS-CoV-2 infection in this setting is still unclear. We documented a 4.8% prevalence of COVID-19 in our study group, remarkably higher than the 0.1% reported in the general population, and median onset of COVID-19 was 169 days after CAR T-cell therapy. Several factors could explain the high rate of COVID-19 in patients receiving CAR T cells. Our study included a homogeneous group of heavily pretreated patients with large B-cell lymphoma who received at least 2 lines of treatment before CAR T-cell and lymphodepletion therapies. Delayed cytopenias and impaired immune reconstitution, leading to a significant risk of infectious complications, have been well documented after CAR T-cell therapy.12  Consistent with findings seen in prior studies, we observed a low lymphocyte count 90 days after CAR T cells (median ALC, 435/mm3) and at the time of COVID-19 diagnosis (median ALC, 370/mm3; 23% of patients had <200 lymphocytes per mm3), although the presence of both neutropenia and lymphocytopenia at COVID-19 diagnosis was not statistically significant in univariable analysis. In addition, it should be emphasized that the presence of relapsed/refractory disease in one-third of patients at the time of COVID-19 should be taken into account as a potential confounding factor. Regrettably, we were unable to investigate in detail humoral or cellular immune reconstitution or whether worsening of lymphopenia during infection had an impact on survival. Notably, our results showed that half of the patients developed COVID-19 after the first 6 months post–CAR T-cell therapy, underscoring that prolonged delayed immune recovery may persist for a long period of time after cellular therapy.17  Currently, there are few clinical trials evaluating the potential role of COVID-19 treatment in patients with cancer18 ; observational studies are also extremely limited. In our study, patients received a wide array of treatments, making difficult to draw any firm conclusions. Based on the presence of impaired humoral and cellular immune reconstitution after CAR T-cell therapy in a consistent number of patients, suboptimal responses to current treatments used in patients with COVID-19 are expected, although specific studies are urgently required to address this issue.

Mortality rates in patients receiving cellular therapy have been reported in few studies. Altuntas et al9  evaluated 32 recipients of autologous and allogeneic grafts and found a 33% case fatality rate among patients still receiving immunosuppressive agents at the time of COVID-19 diagnosis. Similar results have been reported by the European Society for Blood and Marrow Transplantation group, with a mortality rate reaching 25%; older age, need for ICU admission, and moderate/high immunodeficiency index increased the risk for mortality.19  The Center for International Blood and Marrow Transplant Research reported the results of 318 patients with COVID-19 who had undergone HSCT (allogeneic, n = 184; autologous, n = 134). Overall mortality was 22% among those who underwent allogeneic HSCT; age > 50 years, male sex, and time from HSCT to COVID-19 diagnosis <6 months were factors significantly associated with mortality.20  Shah et al10  evaluated 77 patients with COVID-19 who had been treated with HSCT (n = 72) and CAR T-cell therapy (n = 5); the overall death rate was 41%, roughly similar to that reported in our study, and was largely driven by patients with advance disease. The 50% mortality rate was remarkably high in our patients, considering that 10 of 15 deaths resulted from COVID-19 or COVID-19–related events. In this respect, it is worthwhile to keep in mind that two-thirds of our patients had severe infections, and 30% of our patients required mechanical ventilation. In addition, advanced disease status at the time of COVID-19 diagnosis should be considered a potential factor limiting favorable outcomes for patients, as emphasized by the univariable analysis. Several factors might explain the higher mortality rate observed in our study as compared with that reported in patients who had undergone HSCT. Patients with relevant comorbidities are usually excluded from HSCT programs, whereas comorbidities do not preclude CAR T-cell treatment in these vulnerable patients. Time interval from HSCT to COVID-19 has emerged as a factor associated with mortality in many studies, ranging from 13.7 to 18.9 months,12,14,19,20  significantly longer than the median time from CAR T-cell therapy to COVID-19 reported in our series (median time, 169 days).

Preliminary data in patients who have undergone HSCT show that COVID-19 is associated with low lymphocyte counts, particularly in T-cell compartments; however, lymphopenia does not seem to impair immune reconstitution in recovery from COVID-19. Regrettably, we do not have data on lymphocyte subsets after COVID-19 in our cohort of patients, although protracted and profound lymphopenias after CAR T cells have been reported in several studies. In addition, whether differences in CAR T-cell products affect the kinetics of immunodeficiency recovery remains to be determined.

If prospective large studies corroborate our preliminary results, it seems wise to define strategies able to mitigate the adverse events occurring in patients receiving CAR T cells who develop COVID-19. Prioritization of COVID-19 vaccination in patients with hematological disease is of paramount importance; however, based on existing knowledge of the reduced immunogenicity in the immunocompromised host, CAR T-cell recipients are not expected to generate robust responses to COVID-19 vaccines. In this respect, additional preventive measures should be explored. It has been shown that cellular therapies may be safely administered throughout the COVID-19 pandemic when appropriate interventions are instituted, including antimicrobial stewardship programs, screening of donors and recipients, and safe delivery of cellular products. Appealing alternatives to vaccination are monoclonal antibodies or prophylaxis with oral agents (fluvoxamine and molnupiravir), although clinical trials in patients with hematological disease are urgently needed.

Our study is limited by its retrospective nature and small number of patients. Nevertheless, our results highlight a significant mortality rate in patients with COVID-19 who have received CAR T-cell therapy. Therapeutic strategies will need to be developed to ensure that CAR T-cell therapy can be delivered safely and successfully while COVID-19 remains endemic. Furthermore, data on vaccinations in this cohort are eagerly awaited to help formulate safe delivery.

The authors thank all contributors for their utmost contributions and support to the project during a pandemic situation and to Susann Blossfeld and Corinna Kramer for their administrative and technical assistance.

EPICOVIDEHA has received funds from the Optics COMMITTM (COVID-19 Unmet Medical Needs and Associated Research Extension) COVID-19 RFP program by Gilead Sciences (Foster City, CA; project 2020-8223).

Contribution: A.B., J.S.-G., P.C., and L.P. conceived of and codesigned the database; A.B., J.S.-G., and L.P. contributed to data analysis; A.B. wrote the manuscript; and all authors critically reviewed and revised the manuscript and provided final approval.

Conflict-of-interest disclosure: A.B. has received lecture honoraria from Gilead Sciences, Merck, Pfizer Pharmaceuticals, Basilea Pharmaceutica, Biotest, and Jazz Pharmaceuticals and served as a board member of Gilead Sciences and Takeda Pharmaceutical Company. S.L. has received support from Janssen, Gilead Sciences, Roche, AbbVie, Sanofi, Novartis, Actelion, and Pfizer and a research grant from Janssen. L.D. has received lecture honoraria from AbbVie, Amgen, Celgene, Egis, Gilead Sciences, Kyowa Kirin, Merck Sharp & Dohme, Pfizer, Roche, Sandoz, Servier Laboratories, Takeda Pharmaceutical Company, and Teva Pharmaceutical Industries and served as a board member of AbbVie, Gilead Sciences, Novartis, Pfizer, and Takeda Pharmaceutical Company. M.H. reports research funding from Gilead Sciences, Astellas Pharma, Pfizer, and Merck Sharp & Dohme, outside the submitted work. N.K. reports research funding and honoraria from Gilead Sciences, Astellas Pharma, Pfizer, and Merck Sharp & Dohme, outside the submitted work. P.K. is supported by the German Federal Ministry of Research and Education and the State of North Rhine-Westphalia, Germany; has received nonfinancial scientific grants from Miltenyi Biotech GmbH (Bergisch Gladbach, Germany) and the Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne (Cologne, Germany); and has received lecture honoraria from and/or is an advisor to the Akademie für Infektionsmedizin e.V., Ambu GmbH, Astellas Pharma, the European Confederation of Medical Mycology, Gilead Sciences, GPR Academy Ruesselsheim, Merck Sharp & Dohme GmbH, Noxxon N.V., Pfizer Pharma GmbH, and the University Hospital, Ludwig Maximilian University of Munich, outside the submitted work. O.A.C. reports grants or contracts from Amplyx Pharmaceuticals, Basilea Pharmaceutica, the German Federal Ministry of Education and Research, Cidara Therapeutics, the German Centre for Infection Research, the EU Directorate-General for Research and Innovation (101037867), F2G, Gilead Sciences, Matinas BioPharma, Medpace, Merck Sharp & Dohme, Mundipharma, Octapharma, Pfizer, and Scynexis; consulting fees from Amplyx Pharmaceuticals, Biocon, Biosys, Cidara Therapeutics, Da Volterra, Gilead Sciences, Matinas BioPharma, Medpace, Menarini, Molecular Partners, MSGERC, Noxxon Pharma, Octapharma, PSI, Scynexis, and Seres Therapeutics; honoraria for lectures from Abbott Laboratories, Al-Jazeera Pharmaceutical Industries, Astellas Pharma, Grupo Biotoscana/United Medical/Knight, Hikma Pharmaceuticals, Medscape, Medupdate, Merck/Merck Sharp & Dohme, Mylan, and Pfizer; payment for expert testimony from Cidara Therapeutics; participation on a data safety monitoring board or advisory board for Actelion, Allecra Therapeutics, Cidara Therapeutics, Entasis Therapeutics, IQVIA, Janssen, Medpace, Paratek Pharma, PSI, and Shionogi; a pending patent currently being reviewed at the German Patent and Trade Mark Office; and other interests from DGHO, DGI, ECMM, ISHAM, MSGERC, and Wiley. The remaining authors declare no competing financial interests.

A complete list of the members of the EPICOVIDEHA Study Group appears in “ Appendix.”

Correspondence: Alessandro Busca, Stem Cell Transplant Unit, AOU Cittadella Salute e della Scienza, Corso Bramante 88, 10126 Turin, Italy; e-mail: abusca@cittadellasalute.to.it; and Jon Salmanton-García, University Hospital Cologne, Internal Medicine I, Infectious Diseases 2, Herderstrasse 52-54, 50931 Cologne, Germany; e-mail: jon.salmanton-garcia@uk-koeln.de.

Appendix: study group members

Investigators of the EPICOVIDEHA Study Group have 2 different statuses: authors and collaborators. Authors: Alessandro Busca, Jon Salmanton-García, Paolo Corradini, Francesco Marchesi, Alba Cabirta, Roberta Di Blasi, Remy Dulery, Sylvain Lamure, Francesca Farina, Barbora Weinbergerová, Josip Batinić, Anna Nordlander, Alberto López-García, Ľuboš Drgoňa, Ildefonso Espigado-Tocino, Iker Falces-Romero, Ramón García-Sanz, Carolina García-Vidal, Anna Guidetti, Nina Khanna, Austin Kulasekararaj, Johan Maertens, Martin Hoenigl, Nikolai Klimko, Philipp Koehler, Antonio Pagliuca, Francesco Passamonti, Oliver A. Cornely, and Livio Pagano. Collaborators: Jiří Mayer, Cristina de Ramón-Sánchez, Jenna Essame, Nicole García-Poutón, Raúl Córdoba, Marina Machado, Juan-Alberto Martín-González, Ola Blennow, Elisa Roldán, and Jakob Passweg.

1.
Guan
WJ
,
Ni
ZY
,
Hu
Y
, et al;
China Medical Treatment Expert Group for Covid-19
.
Clinical characteristics of coronavirus disease 2019 in China
.
N Engl J Med.
2020
;
382
(
18
):
1708
-
1720
.
2.
Miyashita
H
,
Mikami
T
,
Chopra
N
, et al
.
Do patients with cancer have a poorer prognosis of COVID-19? An experience in New York City
.
Ann Oncol.
2020
;
31
(
8
):
1088
-
1089
.
3.
Passamonti
F
,
Cattaneo
C
,
Arcaini
L
, et al
.
Clinical characteristics and risk factors associated with COVID-19 severity in patients with haematological malignancies in Italy: a retrospective, multicentre, cohort study
.
Lancet Haematol.
2020
;
7
(
10
):
e737
-
e745
.
4.
Mehta
V
,
Goel
S
,
Kabarriti
R
, et al
.
Case fatality rate of cancer patients with COVID-19 in a New York hospital system
.
Cancer Discov.
2020
;
10
(
7
):
935
-
941
.
5.
Malard
F
,
Genthon
A
,
Brissot
E
, et al
.
COVID-19 outcomes in patients with hematologic disease
.
Bone Marrow Transplant.
2020
;
55
(
11
):
2180
-
2184
.
6.
Piñana
JL
,
Martino
R
,
García-García
I
, et al;
Infectious Complications Subcommittee of the Spanish Hematopoietic Stem Cell Transplantation and Cell Therapy Group (GETH)
.
Risk factors and outcome of COVID-19 in patients with hematological malignancies
.
Exp Hematol Oncol.
2020
;
9
:
21
.
7.
Martín-Moro
F
,
Marquet
J
,
Piris
M
, et al
.
Survival study of hospitalised patients with concurrent COVID-19 and haematological malignancies
.
Br J Haematol.
2020
;
190
(
1
):
e16
-
e20
.
8.
Scarfò
L
,
Chatzikonstantinou
T
,
Rigolin
GM
, et al
.
COVID-19 severity and mortality in patients with chronic lymphocytic leukemia: a joint study by ERIC, the European Research Initiative on CLL, and CLL Campus
.
Leukemia.
2020
;
34
(
9
):
2354
-
2363
.
9.
Altuntas
F
,
Ata
N
,
Yigenoglu
TN
, et al;
Turkish Ministry of Health, Hematology Scientific Working Group
.
COVID-19 in hematopoietic cell transplant recipients
.
Bone Marrow Transplant.
2021
;
56
(
4
):
952
-
955
.
10.
Shah
GL
,
DeWolf
S
,
Lee
YJ
, et al
.
Favorable outcomes of COVID-19 in recipients of hematopoietic cell transplantation
.
J Clin Invest.
2020
;
130
(
12
):
6656
-
6667
.
11.
Sterner
RC
,
Sterner
RM
.
CAR-T cell therapy: current limitations and potential strategies
.
Blood Cancer J.
2021
;
11
(
4
):
69
.
12.
Strati
P
,
Varma
A
,
Adkins
S
, et al
.
Hematopoietic recovery and immune reconstitution after axicabtagene ciloleucel in patients with large B-cell lymphoma
.
Haematologica.
2021
;
106
(
10
):
2667
-
2672
.
13.
Salmanton-García
J
,
Busca
A
,
Cornely
OA
, et al
.
EPICOVIDEHA: a ready-to use platform for epidemiological studies in hematological patients with COVID-19
.
HemaSphere.
2021
;
5
(
7
):
e612
.
14.
Shah
V
,
Ko Ko
T
,
Zuckerman
M
, et al
.
Poor outcome and prolonged persistence of SARS-CoV-2 RNA in COVID-19 patients with haematological malignancies; King’s College Hospital experience
.
Br J Haematol.
2020
;
190
(
5
):
e279
-
e282
.
15.
Maia
C
,
Martín-Sánchez
E
,
Garcés
JJ
, et al
.
Immunologic characterization of COVID-19 patients with hematological cancer
.
Haematologica.
2020
;
106
(
5
):
1457
-
1460
.
16.
Vijenthira
A
,
Gong
IY
,
Fox
TA
, et al
.
Outcomes of patients with hematologic malignancies and COVID-19: a systematic review and meta-analysis of 3377 patients
.
Blood.
2020
;
136
(
25
):
2881
-
2892
.
17.
Logue
JM
,
Zucchetti
E
,
Bachmeier
CA
, et al
.
Immune reconstitution and associated infections following axicabtagene ciloleucel in relapsed or refractory large B-cell lymphoma
.
Haematologica.
2020
;
106
(
4
):
978
-
986
.
18.
Rivera
DR
,
Peters
S
,
Panagiotou
OA
, et al
.
Utilization of COVID-19 treatments and clinical outcomes among patients with cancer: a COVID-19 and cancer consortium (CCC19) cohort study
.
Cancer Discov.
2020
;
10
(
10
):
1514
-
1527
.
19.
Ljungman
P
,
de la Camara
R
,
Mikulska
M
, et al
.
COVID-19 and stem cell transplantation; results from an EBMT and GETH multicenter prospective survey
.
Leukemia.
2021
;
35
(
10
):
2885
-
2894
.
20.
Sharma
A
,
Bhatt
NS
,
St Martin
A
, et al
.
Clinical characteristics and outcomes of COVID-19 in haematopoietic stem-cell transplantation recipients: an observational cohort study [published correction appears in Lancet Haematol. 2021;8(6):e393]
.
Lancet Haematol.
2021
;
8
(
3
):
e185
-
e193
.

Author notes

*

A.B. and J.S.-G. contributed equally to this study.

Requests for data sharing may be submitted to Alessandro Busca (abusca@cittadellasalute.to.it).